1
|
Wieg L, Ciola JC, Wasén CC, Gaba F, Colletti BR, Schroeder MK, Hinshaw RG, Ekwudo MN, Holtzman DM, Saito T, Sasaguri H, Saido TC, Cox LM, Lemere CA. Cognitive Effects of Simulated Galactic Cosmic Radiation Are Mediated by ApoE Status, Sex, and Environment in APP Knock-In Mice. Int J Mol Sci 2024; 25:9379. [PMID: 39273325 PMCID: PMC11394682 DOI: 10.3390/ijms25179379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/25/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
Cosmic radiation experienced during space travel may increase the risk of cognitive impairment. While simulated galactic cosmic radiation (GCRsim) has led to memory deficits in wildtype (WT) mice, it has not been investigated whether GCRsim in combination with genetic risk factors for Alzheimer's disease (AD) worsens memory further in aging mice. Here, we investigated the central nervous system (CNS) effects of 0 Gy (sham) or 0.75 Gy five-ion GCRsim or 2 Gy gamma radiation (IRR) in 14-month-old female and male APPNL-F/NL-F knock-in (KI) mice bearing humanized ApoE3 or ApoE4 (APP;E3F and APP;E4F). As travel to a specialized facility was required for irradiation, both traveled sham-irradiated C57BL/6J WT and KI mice and non-traveled (NT) KI mice acted as controls for potential effects of travel. Mice underwent four behavioral tests at 20 months of age and were euthanized for pathological and biochemical analyses 1 month later. Fecal samples were collected pre- and post-irradiation at four different time points. GCRsim seemed to impair memory in male APP;E3F mice compared to their sham counterparts. Travel tended to improve cognition in male APP;E3F mice and lowered total Aβ in female and male APP;E3F mice compared to their non-traveled counterparts. Sham-irradiated male APP;E4F mice accumulated more fibrillar amyloid than their APP;E3F counterparts. Radiation exposure had only modest effects on behavior and brain changes, but travel-, sex-, and genotype-specific effects were seen. Irradiated mice had immediate and long-term differences in their gut bacterial composition that correlated to Alzheimer's disease phenotypes.
Collapse
Affiliation(s)
- Laura Wieg
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Jason C. Ciola
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Caroline C. Wasén
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Fidelia Gaba
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Brianna R. Colletti
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Maren K. Schroeder
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - Robert G. Hinshaw
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Millicent N. Ekwudo
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
| | - David M. Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer’s Disease Research Center, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Takashi Saito
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya 467-8601, Aichi, Japan;
| | - Hiroki Sasaguri
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan; (H.S.); (T.C.S.)
| | - Takaomi C. Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako City 351-0198, Saitama, Japan; (H.S.); (T.C.S.)
| | - Laura M. Cox
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| | - Cynthia A. Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Boston, MA 02115, USA; (L.W.); (J.C.C.); (C.C.W.); (F.G.); (B.R.C.); (M.K.S.); (R.G.H.); (M.N.E.); (L.M.C.)
- Department of Neurology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
2
|
van Heuvelen MJG, van der Lei MB, Alferink PM, Roemers P, van der Zee EA. Cognitive deficits in human ApoE4 knock-in mice: A systematic review and meta-analysis. Behav Brain Res 2024; 471:115123. [PMID: 38972485 DOI: 10.1016/j.bbr.2024.115123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 07/09/2024]
Abstract
Apolipoprotein-E4 (ApoE4) is an important genetic risk factor for Alzheimer's disease. The development of targeted-replacement human ApoE knock-in mice facilitates research into mechanisms by which ApoE4 affects the brain. We performed meta-analyses and meta-regression analyses to examine differences in cognitive performance between ApoE4 and ApoE3 mice. We included 61 studies in which at least one of the following tests was assessed: Morris Water Maze (MWM), novel object location (NL), novel object recognition (NO) and Fear Conditioning (FC) test. ApoE4 vs. ApoE3 mice performed significantly worse on the MWM (several outcomes, 0.17 ≤ g ≤ 0.60), NO (exploration, g=0.33; index, g=0.44) and FC (contextual, g=0.49). ApoE4 vs. ApoE3 differences were not systematically related to sex or age. We conclude that ApoE4 knock-in mice in a non-AD condition show some, but limited cognitive deficits, regardless of sex and age. These effects suggest an intrinsic vulnerability in ApoE4 mice that may become more pronounced under additional brain load, as seen in neurodegenerative diseases.
Collapse
Affiliation(s)
- Marieke J G van Heuvelen
- Department of Human Movement Sciences, University of Groningen, University Medical Center Groningen, Groningen, A. Deusinglaan 1, Groningen 9713AV, the Netherlands.
| | - Mathijs B van der Lei
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborg 7, Groningen 9747 AG, the Netherlands; Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, Prins Boudewijnlaan 43, Edegem 2650, Belgium.
| | - Pien M Alferink
- Department of Human Movement Sciences, University of Groningen, University Medical Center Groningen, Groningen, A. Deusinglaan 1, Groningen 9713AV, the Netherlands.
| | - Peter Roemers
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborg 7, Groningen 9747 AG, the Netherlands.
| | - Eddy A van der Zee
- Molecular Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborg 7, Groningen 9747 AG, the Netherlands.
| |
Collapse
|
3
|
Wang W, Zhao F, Torres S, Harris PL, Wang X, Peng L, Siedlak SL, Zhu X. Space-Like Irradiation Exacerbated Cognitive Deficits and Amyloid Pathology in CRND8 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2024; 100:S327-S339. [PMID: 39058444 PMCID: PMC11875098 DOI: 10.3233/jad-240570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Background Space radiation was linked to neurological damage and behavioral deficits which raised concerns of increased degenerative risk on the brain and development of Alzheimer's disease following space travel. Objective In this study, we investigated the effects of irradiation by 56Fe and 28Si in CRND8 mice, an Alzheimer's disease mouse model. Methods Six-month-old CRND8 mice were exposed to whole body irradiation by 56Fe and 28Si at 0.5 Gy and 2 Gy doses. Behavior tests were administered 1-month to 3-months post-irradiation. Amyloid deposition and other pathological changes were analyzed 3-months and/or 6-months post-irradiation. Results The Novel Object Recognition test showed some decline in 8-month-old mice compared to non-irradiated CRND8 mice. Male mice also showed a loss of freezing behavior in the fear conditioning contextual test following irradiation. Golgi staining revealed a loss of spines in hippocampal neurons after irradiation. Total amyloid immunohistochemistry showed a robust increase in 3-months post-irradiation 56Fe groups which became normalized to non-irradiated group by 6-months post-irradiation. However, 2 Gy 28Si caused a trend towards increased plaque load at 3-months post-irradiation which became significant at 6-months post irradiation only in male CRND8 mice. While 0.5 Gy Fe did not induce obvious changes in the total number of iba-1 positive microglia, more hippocampal microglia were found to express PCNA after 0.5 Gy Fe treatment, suggesting potential involvement of microglial dysfunction. Conclusions Overall, our study provides new evidence of gender-specific and ion-dependent effects of space radiation on cognition and amyloid pathology in AD models.
Collapse
Affiliation(s)
- Wenzhang Wang
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Fanpeng Zhao
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sandy Torres
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Peggy L.R. Harris
- Department of Genetics, Case Western Reserve University, Cleveland, OH, USA
| | - Xinglong Wang
- Department of Pharmacology & Toxicology, University of Arizona, Tucson, AZ, USA
| | - Lihua Peng
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Sandra L. Siedlak
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
4
|
Kundu P, Zimmerman B, Quinn JF, Kaye J, Mattek N, Westaway SK, Raber J. Serum Levels of α-Klotho Are Correlated with Cerebrospinal Fluid Levels and Predict Measures of Cognitive Function. J Alzheimers Dis 2022; 86:1471-1481. [PMID: 35213382 PMCID: PMC9108571 DOI: 10.3233/jad-215719] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 02/05/2023]
Abstract
BACKGROUND α-klotho might play a role in neurodegenerative diseases. OBJECTIVE To determine levels of α-klotho and apoE in serum and cerebrospinal fluid (CSF) samples and their relationship with the Mini-Mental State Examination (MMSE) and Clinical Dementia Rating (CDR). METHODS All subjects were between age 39 to 83+ (n = 94). CDR and MMSE were administered to all participants. CSF was collected in the early afternoon by lumbar puncture. RESULTS Serum and CSF levels of α-klotho are positively correlated and both predict scores on the MMSE and CDR, regardless of sex or apoE4 status. CONCLUSION Our results demonstrate that α-klotho may be an important biomarker of cognitive health and neurodegeneration, and that relatively non-invasive sampling of α-klotho from serum is likely highly reflective of CSF levels.
Collapse
Affiliation(s)
- Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Veterans Affairs Medical Center, Portland, OR, USA
| | - Jeffrey Kaye
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora Mattek
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Shawn K. Westaway
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
- Departments of Psychiatry and Radiation Medicine, Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
5
|
Long-Term Sex- and Genotype-Specific Effects of 56Fe Irradiation on Wild-Type and APPswe/PS1dE9 Transgenic Mice. Int J Mol Sci 2021; 22:ijms222413305. [PMID: 34948098 PMCID: PMC8703695 DOI: 10.3390/ijms222413305] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
Space radiation presents a substantial threat to travel beyond Earth. Relatively low doses of high-energy particle radiation cause physiological and behavioral impairments in rodents and may pose risks to human spaceflight. There is evidence that 56Fe irradiation, a significant component of space radiation, may be more harmful to males than to females and worsen Alzheimer's disease pathology in genetically vulnerable models. Yet, research on the long-term, sex- and genotype-specific effects of 56Fe irradiation is lacking. Here, we irradiated 4-month-old male and female, wild-type and Alzheimer's-like APP/PS1 mice with 0, 0.10, or 0.50 Gy of 56Fe ions (1GeV/u). Mice underwent microPET scans before and 7.5 months after irradiation, a battery of behavioral tests at 11 months of age and were sacrificed for pathological and biochemical analyses at 12 months of age. 56Fe irradiation worsened amyloid-beta (Aβ) pathology, gliosis, neuroinflammation and spatial memory, but improved motor coordination, in male transgenic mice and worsened fear memory in wild-type males. Although sham-irradiated female APP/PS1 mice had more cerebral Aβ and gliosis than sham-irradiated male transgenics, female mice of both genotypes were relatively spared from radiation effects 8 months later. These results provide evidence for sex-specific, long-term CNS effects of space radiation.
Collapse
|
6
|
Kundu P, Zimmerman B, Perez R, Whitlow CT, Cline JM, Olson JD, Andrews RN, Raber J. Apolipoprotein E levels in the amygdala and prefrontal cortex predict relative regional brain volumes in irradiated Rhesus macaques. Sci Rep 2021; 11:22130. [PMID: 34764354 PMCID: PMC8585884 DOI: 10.1038/s41598-021-01480-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 10/26/2021] [Indexed: 01/20/2023] Open
Abstract
In the brain, apolipoprotein E (apoE) plays an important role in lipid transport and response to environmental and age-related challenges, including neuronal repair following injury. While much has been learned from radiation studies in rodents, a gap in our knowledge is how radiation might affect the brain in primates. This is important for assessing risk to the brain following radiotherapy as part of cancer treatment or environmental radiation exposure as part of a nuclear accident, bioterrorism, or a nuclear attack. In this study, we investigated the effects of ionizing radiation on brain volumes and apoE levels in the prefrontal cortex, amygdala, and hippocampus of Rhesus macaques that were part of the Nonhuman Primate Radiation Survivor Cohort at the Wake Forest University. This unique cohort is composed of Rhesus macaques that had previously received single total body doses of 6.5-8.05 Gy of ionizing radiation. Regional apoE levels predicted regional volume in the amygdala and the prefrontal cortex. In addition, apoE levels in the amygdala, but not the hippocampus, strongly predicted relative hippocampal volume. Finally, radiation dose negatively affected relative hippocampal volume when apoE levels in the amygdala were controlled for, suggesting a protective compensatory role of regional apoE levels following radiation exposure. In a supplementary analysis, there also was a robust positive relationship between the neuroprotective protein α-klotho and apoE levels in the amygdala, further supporting the potentially protective role of apoE. Increased understanding of the effects of IR in the primate brain and the role of apoE in the irradiated brain could inform future therapies to mitigate the adverse effects of IR on the CNS.
Collapse
Affiliation(s)
- Payel Kundu
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Benjamin Zimmerman
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
- Advanced Imaging Research Center, Oregon Health and Science University, Portland, OR, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Ruby Perez
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA
| | - Christopher T Whitlow
- Department of Radiology, Radiology Informatics & Image Processing Laboratory (RIIPL), Wake Forest University, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - J Mark Cline
- Department of Radiology, Radiology Informatics & Image Processing Laboratory (RIIPL), Wake Forest University, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - John D Olson
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Rachel N Andrews
- Department of Radiology, Radiology Informatics & Image Processing Laboratory (RIIPL), Wake Forest University, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Department of Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, USA.
- Division of Neuroscience, Departments of Neurology and Radiation Medicine, ONPRC, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
7
|
Soler I, Yun S, Reynolds RP, Whoolery CW, Tran FH, Kumar PL, Rong Y, DeSalle MJ, Gibson AD, Stowe AM, Kiffer FC, Eisch AJ. Multi-Domain Touchscreen-Based Cognitive Assessment of C57BL/6J Female Mice Shows Whole-Body Exposure to 56Fe Particle Space Radiation in Maturity Improves Discrimination Learning Yet Impairs Stimulus-Response Rule-Based Habit Learning. Front Behav Neurosci 2021; 15:722780. [PMID: 34707486 PMCID: PMC8543003 DOI: 10.3389/fnbeh.2021.722780] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 09/08/2021] [Indexed: 12/23/2022] Open
Abstract
Astronauts during interplanetary missions will be exposed to galactic cosmic radiation, including charged particles like 56Fe. Most preclinical studies with mature, "astronaut-aged" rodents suggest space radiation diminishes performance in classical hippocampal- and prefrontal cortex-dependent tasks. However, a rodent cognitive touchscreen battery unexpectedly revealed 56Fe radiation improves the performance of C57BL/6J male mice in a hippocampal-dependent task (discrimination learning) without changing performance in a striatal-dependent task (rule-based learning). As there are conflicting results on whether the female rodent brain is preferentially injured by or resistant to charged particle exposure, and as the proportion of female vs. male astronauts is increasing, further study on how charged particles influence the touchscreen cognitive performance of female mice is warranted. We hypothesized that, similar to mature male mice, mature female C57BL/6J mice exposed to fractionated whole-body 56Fe irradiation (3 × 6.7cGy 56Fe over 5 days, 600 MeV/n) would improve performance vs. Sham conditions in touchscreen tasks relevant to hippocampal and prefrontal cortical function [e.g., location discrimination reversal (LDR) and extinction, respectively]. In LDR, 56Fe female mice more accurately discriminated two discrete conditioned stimuli relative to Sham mice, suggesting improved hippocampal function. However, 56Fe and Sham female mice acquired a new simple stimulus-response behavior and extinguished this acquired behavior at similar rates, suggesting similar prefrontal cortical function. Based on prior work on multiple memory systems, we next tested whether improved hippocampal-dependent function (discrimination learning) came at the expense of striatal stimulus-response rule-based habit learning (visuomotor conditional learning). Interestingly, 56Fe female mice took more days to reach criteria in this striatal-dependent rule-based test relative to Sham mice. Together, our data support the idea of competition between memory systems, as an 56Fe-induced decrease in striatal-based learning is associated with enhanced hippocampal-based learning. These data emphasize the power of using a touchscreen-based battery to advance our understanding of the effects of space radiation on mission critical cognitive function in females, and underscore the importance of preclinical space radiation risk studies measuring multiple cognitive processes, thereby preventing NASA's risk assessments from being based on a single cognitive domain.
Collapse
Affiliation(s)
- Ivan Soler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sanghee Yun
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ryan P. Reynolds
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Cody W. Whoolery
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Fionya H. Tran
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Priya L. Kumar
- University of Pennsylvania, Philadelphia, PA, United States
| | - Yuying Rong
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Matthew J. DeSalle
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Adam D. Gibson
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ann M. Stowe
- Department of Neurology and Neurological Therapeutics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Frederico C. Kiffer
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Amelia J. Eisch
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Department of Anesthesiology and Critical Care Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neuroscience, Perelman School of Medicine, Mahoney Institute for Neurosciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
8
|
Minnier J, Emmett MR, Perez R, Ding LH, Barnette BL, Larios RE, Hong C, Hwang TH, Yu Y, Fallgren CM, Story MD, Weil MM, Raber J. Associations between lipids in selected brain regions, plasma miRNA, and behavioral and cognitive measures following 28Si ion irradiation. Sci Rep 2021; 11:14899. [PMID: 34290258 PMCID: PMC8295277 DOI: 10.1038/s41598-021-93869-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 06/29/2021] [Indexed: 11/09/2022] Open
Abstract
The space radiation environment consists of multiple species of charged particles, including 28Si ions, that may impact brain function during and following missions. To develop biomarkers of the space radiation response, BALB/c and C3H female and male mice and their F2 hybrid progeny were irradiated with 28Si ions (350 MeV/n, 0.2 Gy) and tested for behavioral and cognitive performance 1, 6, and 12 months following irradiation. The plasma of the mice was collected for analysis of miRNA levels. Select pertinent brain regions were dissected for lipidomic analyses and analyses of levels of select biomarkers shown to be sensitive to effects of space radiation in previous studies. There were associations between lipids in select brain regions, plasma miRNA, and cognitive measures and behavioral following 28Si ion irradiation. Different but overlapping sets of miRNAs in plasma were found to be associated with cognitive measures and behavioral in sham and irradiated mice at the three time points. The radiation condition revealed pathways involved in neurodegenerative conditions and cancers. Levels of the dendritic marker MAP2 in the cortex were higher in irradiated than sham-irradiated mice at middle age, which might be part of a compensatory response. Relationships were also revealed with CD68 in miRNAs in an anatomical distinct fashion, suggesting that distinct miRNAs modulate neuroinflammation in different brain regions. The associations between lipids in selected brain regions, plasma miRNA, and behavioral and cognitive measures following 28Si ion irradiation could be used for the development of biomarker of the space radiation response.
Collapse
Affiliation(s)
- Jessica Minnier
- Oregon Health & Science University-Portland State University School of Public Health, Knight Cancer Institute Biostatistics Shared Resource, and the Knight Cardiovascular Institute, OR Health & Science University, Portland, OR, 97239, USA
| | - Mark R Emmett
- Department of Biochemistry and Molecular Biology; Radiation Oncology, Pharmacology and Toxicology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch Cancer Center, Galveston, TX, 77555, USA
| | - Ruby Perez
- Department of Behavioral Neuroscience, L470, Oregon Health & Science University, 3181SW Sam Jackson Park Road, Portland, OR, 97239, USA
| | - Liang-Hao Ding
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Brooke L Barnette
- Department of Biochemistry and Molecular Biology; Radiation Oncology, Pharmacology and Toxicology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch Cancer Center, Galveston, TX, 77555, USA
| | - Rianna E Larios
- Department of Biochemistry and Molecular Biology; Radiation Oncology, Pharmacology and Toxicology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch Cancer Center, Galveston, TX, 77555, USA
| | - Changjin Hong
- Lerner Research Institute, Cleveland Clinic Lerner College of Medicine US, Cleveland, OH, 44195, USA
| | - Tae Hyun Hwang
- Lerner Research Institute, Cleveland Clinic Lerner College of Medicine US, Cleveland, OH, 44195, USA
- Department of Molecular Medicine, School of Medicine, GU Malignancies Program, Case Comprehensive Cancer Center, Genomic Medicine Institute, Case Western Reserve University US., Cleveland, OH, 10900, USA
| | - Yongjia Yu
- Department of Biochemistry and Molecular Biology; Radiation Oncology, Pharmacology and Toxicology, Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch Cancer Center, Galveston, TX, 77555, USA
| | - Christina M Fallgren
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Michael D Story
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael M Weil
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, L470, Oregon Health & Science University, 3181SW Sam Jackson Park Road, Portland, OR, 97239, USA.
- Division of Neuroscience ONPRC, Departments of Neurology, Psychiatry, and Radiation Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
| |
Collapse
|
9
|
Boerma M, Davis CM, Jackson IL, Schaue D, Williams JP. All for one, though not one for all: team players in normal tissue radiobiology. Int J Radiat Biol 2021; 98:346-366. [PMID: 34129427 PMCID: PMC8781287 DOI: 10.1080/09553002.2021.1941383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE As part of the special issue on 'Women in Science', this review offers a perspective on past and ongoing work in the field of normal (non-cancer) tissue radiation biology, highlighting the work of many of the leading contributors to this field of research. We discuss some of the hypotheses that have guided investigations, with a focus on some of the critical organs considered dose-limiting with respect to radiation therapy, and speculate on where the field needs to go in the future. CONCLUSIONS The scope of work that makes up normal tissue radiation biology has and continues to play a pivotal role in the radiation sciences, ensuring the most effective application of radiation in imaging and therapy, as well as contributing to radiation protection efforts. However, despite the proven historical value of preclinical findings, recent decades have seen clinical practice move ahead with altered fractionation scheduling based on empirical observations, with little to no (or even negative) supporting scientific data. Given our current appreciation of the complexity of normal tissue radiation responses and their temporal variability, with tissue- and/or organ-specific mechanisms that include intra-, inter- and extracellular messaging, as well as contributions from systemic compartments, such as the immune system, the need to maintain a positive therapeutic ratio has never been more urgent. Importantly, mitigation and treatment strategies, whether for the clinic, emergency use following accidental or deliberate releases, or reducing occupational risk, will likely require multi-targeted approaches that involve both local and systemic intervention. From our personal perspective as five 'Women in Science', we would like to acknowledge and applaud the role that many female scientists have played in this field. We stand on the shoulders of those who have gone before, some of whom are fellow contributors to this special issue.
Collapse
Affiliation(s)
- Marjan Boerma
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Catherine M. Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Isabel L. Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dörthe Schaue
- Department of Radiation Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
10
|
Liu RM, Chong Z, Chen JC. Ozone and Particulate Matter Exposure and Alzheimer's Disease: A Review of Human and Animal Studies. J Alzheimers Dis 2021; 76:807-824. [PMID: 32568209 DOI: 10.3233/jad-200435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD), an aging-related neurodegenerative disease, is a major cause of dementia in the elderly. Although the early-onset (familial) AD is attributed to mutations in the genes coding for amyloid-β protein precursor (AβPP) and presenilin1/presenilin 2 (PS1/PS2), the cause for the late-onset AD (LOAD), which accounts for more than 95% of AD cases, remains unclear. Aging is the greatest risk factor for LOAD, whereas the apolipo protein E4 allele (APOEɛ4) is believed to be a major genetic risk factor in acquiring LOAD, with female APOEɛ4 carriers at highest risk. Nonetheless, not all the elderly, even older female APOEɛ4 carriers, develop LOAD, suggesting that other factors, including environmental exposure, must play a role. This review summarizes recent studies that show a potential role of environmental exposure, especially ozone and particulate matter exposure, in the development of AD. Interactions between environmental exposure, genetic risk factor (APOEɛ4), and sex in AD pathophysiology are also discussed briefly. Identification of environmental risk factor(s) and elucidation of the complex interactions between genetic and environmental risk factors plus aging and female sex in the onset of AD will be a key to our understanding of the etiology and pathogenesis of AD and the development of the strategies for its prevention and treatment.
Collapse
Affiliation(s)
- Rui-Ming Liu
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zechen Chong
- Department of Genetics, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jiu-Chiuan Chen
- Department of Biostatistics and Data Science, The University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
11
|
Butterbrod E, Sitskoorn M, Bakker M, Jakobs B, Fleischeuer R, Roijers J, Rutten G, Gehring K. The APOE ε4 allele in relation to pre- and postsurgical cognitive functioning of patients with primary brain tumors. Eur J Neurol 2021; 28:1665-1676. [PMID: 33342004 PMCID: PMC8247965 DOI: 10.1111/ene.14693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Recent studies suggest a relationship between the APOE ε4 allele and cognitive outcome in patients treated for malignant brain tumors. Still, longitudinal investigations that include a pretreatment cognitive assessment are lacking and APOE's effects in patients with benign tumors are understudied. This study investigated presurgical cognitive performance and postsurgical change in ε4-carrying and non-carrying patients with glioma and meningioma. METHODS Neuropsychological test scores (CNS Vital Signs battery [seven measures], Digit Span Forward/Backward, Letter Fluency test) were obtained as part of a prospective study in which patients with meningioma and glioma underwent cognitive assessment 1 day before (T0, n = 505) and 3 (T3, n = 418) and 12 months after (T12, n = 167) surgery. APOE isoforms were identified retrospectively. ε4 carriers and non-carriers were compared with regard to pretreatment cognitive performance on the group and individual level. Changes in performances over time were compared with longitudinal mixed model analysis in the total sample and the subgroup receiving adjuvant treatment. RESULTS Carriers and non-carriers did not differ with regard to pretreatment performance. No significant main effect of ε4 carrier status or interaction between time (T0-T12) and carrier status was found on any of the tests in the whole sample nor in the sample receiving adjuvant treatment. CONCLUSIONS This study found no evidence of increased vulnerability for pretreatment cognitive dysfunction or cognitive decline within 1 year after surgery in APOE ε4-carrying meningioma and glioma patients. Investigations that include larger samples at longer-term follow-up are recommended to investigate potential late treatment effects.
Collapse
Affiliation(s)
- Elke Butterbrod
- Department of Cognitive NeuropsychologyTilburg UniversityTilburgThe Netherlands
| | - Margriet Sitskoorn
- Department of Cognitive NeuropsychologyTilburg UniversityTilburgThe Netherlands
| | - Marjan Bakker
- Department of Methodology and StatisticsTilburg UniversityTilburgThe Netherlands
| | - Bernadette Jakobs
- Department of Laboratory MedicineElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Ruth Fleischeuer
- Clinical Pathology LaboratoryElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Janine Roijers
- Department of Laboratory MedicineElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Geert‐Jan Rutten
- Department of NeurosurgeryElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Karin Gehring
- Department of Cognitive NeuropsychologyTilburg UniversityTilburgThe Netherlands
- Department of NeurosurgeryElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| |
Collapse
|
12
|
Lamoureux L, Marottoli FM, Tseng KY, Tai LM. APOE4 Promotes Tonic-Clonic Seizures, an Effect Modified by Familial Alzheimer's Disease Mutations. Front Cell Dev Biol 2021; 9:656521. [PMID: 33796539 PMCID: PMC8007905 DOI: 10.3389/fcell.2021.656521] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/23/2021] [Indexed: 11/13/2022] Open
Abstract
Seizures are emerging as a common symptom in Alzheimer's disease (AD) patients, often attributed to high levels of amyloid β (Aβ). However, the extent that AD disease risk factors modulate seizure activity in aging and AD-relevant contexts is unclear. APOE4 is the greatest genetic risk factor for AD and has been linked to seizures independent of AD and Aβ. The goal of the present study was to evaluate the role of APOE genotype in modulating seizures in the absence and presence of high Aβ levels in vivo. To achieve this goal, we utilized EFAD mice, which express human APOE3 or APOE4 in the absence (EFAD-) or presence (EFAD+) of familial AD mutations that result in Aβ overproduction. When quantified during cage change day, we found that unlike APOE3, APOE4 is associated with tonic-clonic seizures. Interestingly, there were lower tonic-clonic seizures in E4FAD+ mice compared to E4FAD- mice. Restraint handing and auditory stimuli failed to recapitulate the tonic-clonic phenotype in EFAD mice that express APOE4. However, after chemical-induction with pentylenetetrazole, there was a higher incidence of tonic-clonic seizures with APOE4 compared to APOE3. Interestingly, the distribution of seizures to the tonic-clonic phenotype was higher with FAD mutations. These data support that APOE4 is associated with higher tonic-clonic seizures in vivo, and that FAD mutations impact tonic-clonic seizures in a paradigm dependent manner.
Collapse
Affiliation(s)
- Lorissa Lamoureux
- Biological Resources Laboratory, University of Illinois at Chicago, Chicago, IL, United States
| | - Felecia M Marottoli
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Kuei Y Tseng
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M Tai
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
13
|
Raber J, Fuentes Anaya A, Torres ERS, Lee J, Boutros S, Grygoryev D, Hammer A, Kasschau KD, Sharpton TJ, Turker MS, Kronenberg A. Effects of Six Sequential Charged Particle Beams on Behavioral and Cognitive Performance in B6D2F1 Female and Male Mice. Front Physiol 2020; 11:959. [PMID: 32982769 PMCID: PMC7485338 DOI: 10.3389/fphys.2020.00959] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022] Open
Abstract
The radiation environment astronauts are exposed to in deep space includes galactic cosmic radiation (GCR) with different proportions of all naturally occurring ions. To assist NASA with assessment of risk to the brain following exposure to a mixture of ions broadly representative of the GCR, we assessed the behavioral and cognitive performance of female and male C57BL/6J × DBA2/J F1 (B6D2F1) mice two months following rapidly delivered, sequential 6 beam irradiation with protons (1 GeV, LET = 0.24 keV, 50%), 4He ions (250 MeV/n, LET = 1.6 keV/μm, 20%), 16O ions (250 MeV/n, LET = 25 keV/μm 7.5%), 28Si ions (263 MeV/n, LET = 78 keV/μm, 7.5%), 48Ti ions (1 GeV/n, LET = 107 keV/μm, 7.5%), and 56Fe ions (1 GeV/n, LET = 151 keV/μm, 7.5%) at 0, 25, 50, or 200 cGy) at 4-6 months of age. When the activity over 3 days of open field habituation was analyzed in female mice, those irradiated with 50 cGy moved less and spent less time in the center than sham-irradiated mice. Sham-irradiated female mice and those irradiated with 25 cGy showed object recognition. However, female mice exposed to 50 or 200 cGy did not show object recognition. When fear memory was assessed in passive avoidance tests, sham-irradiated mice and mice irradiated with 25 cGy showed memory retention while mice exposed to 50 or 200 cGy did not. The effects of radiation passive avoidance memory retention were not sex-dependent. There was no effect of radiation on depressive-like behavior in the forced swim test. There was a trend toward an effect of radiation on BDNF levels in the cortex of males, but not for females, with higher levels in male mice irradiated with 50 cGy than sham-irradiated. Finally, sequential 6-ion irradiation impacted the composition of the gut microbiome in a sex-dependent fashion. Taxa were uncovered whose relative abundance in the gut was associated with the radiation dose received. Thus, exposure to sequential six-beam irradiation significantly affects behavioral and cognitive performance and the gut microbiome.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
- Departments of Neurology and Radiation Medicine, Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR, United States
| | - Andrea Fuentes Anaya
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Eileen Ruth S. Torres
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Joanne Lee
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Sydney Boutros
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Dmytro Grygoryev
- Oregon Institute of Occupational Health Sciences and Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, United States
| | - Austin Hammer
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Kristin D. Kasschau
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
| | - Thomas J. Sharpton
- Department of Microbiology, Oregon State University, Corvallis, OR, United States
- Department of Statistics, Oregon State University, Corvallis, OR, United States
| | - Mitchell S. Turker
- Oregon Institute of Occupational Health Sciences and Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, United States
| | - Amy Kronenberg
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| |
Collapse
|
14
|
Zaldua S, Damen FC, Pisharody R, Thomas R, Fan KD, Ekkurthi GK, Scheinman SB, Alahmadi S, Marottoli FM, Alford S, Cai K, Tai LM. Epidermal growth factor treatment of female mice that express APOE4 at an age of advanced pathology mitigates behavioral and cerebrovascular dysfunction. Heliyon 2020; 6:e03919. [PMID: 32478184 PMCID: PMC7251379 DOI: 10.1016/j.heliyon.2020.e03919] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/18/2019] [Accepted: 04/30/2020] [Indexed: 02/01/2023] Open
Abstract
APOE4 is a major genetic risk factor for Alzheimer's disease and high amyloid-β (Aβ) levels in the brain are a pathological hallmark of the disease. However, the contribution of specific APOE-modulated Aβ-dependent and Aβ-independent functions to cognitive decline remain unclear. Increasing evidence supports a role of APOE in modulating cerebrovascular function, however whether ameliorating this dysfunction can improve behavioral function is still under debate. We have previously demonstrated that systemic epidermal growth factor (EGF) treatment, which is important for vascular function, at early stages of pathology (treatment from 6 to 8 months) is beneficial for recognition and spatial memory and cerebrovascular function in female mice that express APOE4. These data raise the important question of whether EGF can improve APOE4-associated cerebrovascular and behavioral dysfunction when treatment is initiated at an age of advanced pathology. Positive findings would support the development of therapies that target cerebrovascular dysfunction associated with APOE4 in aging and AD in individuals with advanced cognitive impairment. Therefore, in this study female mice that express APOE4 in the absence (E4FAD- mice) or presence (E4FAD+ mice) of Aβ overproduction were treated from 8 to 10 months of age systemically with EGF. EGF treatment mitigated behavioral dysfunction in recognition memory and spatial learning and improved hippocampal neuronal function in both E4FAD+ and E4FAD- mice, suggesting that EGF treatment improves Aβ-independent APOE4-associated deficits. The beneficial effects of EGF treatment on behavior occurred in tandem with improved markers of cerebrovascular function, including lower levels of fibrinogen, lower permeability when assessed by MRI and higher percent area coverage of laminin and CD31 in the hippocampus. These data suggest a mechanistic link among EGF signaling, cerebrovascular function and APOE4-associated behavioral deficits in mice with advanced AD-relevant pathology.
Collapse
Affiliation(s)
- Steve Zaldua
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Frederick C Damen
- Department of Radiology, University of Illinois Hospital & Health Sciences System, Suite 103, 2242 West Harrison Street, Chicago, IL 60612, USA
| | - Rohan Pisharody
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kelly D Fan
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Giri K Ekkurthi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sarah B Scheinman
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Sami Alahmadi
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Felecia M Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Simon Alford
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Kejia Cai
- Department of Radiology, University of Illinois Hospital & Health Sciences System, Suite 103, 2242 West Harrison Street, Chicago, IL 60612, USA
- Department of Bioengineering, University of Illinois Hospital & Health Sciences System, Suite 103, 2242 West Harrison Street, Chicago, IL 60612, USA
| | - Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
15
|
Pipová Kokošová N, Kisková T, Vilhanová K, Štafuriková A, Jendželovský R, Račeková E, Šmajda B. Melatonin mitigates hippocampal and cognitive impairments caused by prenatal irradiation. Eur J Neurosci 2020; 52:3575-3594. [PMID: 31985866 DOI: 10.1111/ejn.14687] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 12/11/2022]
Abstract
Formation of new neurons and glial cells in the brain is taking place in mammals not only during prenatal embryogenesis but also during adult life. As an enhancer of oxidative stress, ionizing radiation represents a potent inhibitor of neurogenesis and gliogenesis in the brain. It is known that the pineal hormone melatonin is a potent free radical scavenger and counteracts inflammation and apoptosis in brain injuries. The aim of our study was to establish the effects of melatonin on cells in the hippocampus and selected forms of behaviour in prenatally irradiated rats. The male progeny of irradiated (1 Gy of gamma rays; n = 38) and sham-irradiated mothers (n = 19), aged 3 weeks or 2 months, were used in the experiment. Melatonin was administered daily in drinking water (4 mg/kg b. w.) to a subset of animals from each age group. Prenatal irradiation markedly suppressed proliferative activity in the dentate gyrus in both age groups. Melatonin significantly increased the number of proliferative BrdU-positive cells in hilus of young irradiated animals, and the number of mature NeuN-positive neurons in hilus and granular cell layer of the dentate gyrus in these rats and in CA1 region of adult irradiated rats. Moreover, melatonin significantly improved the spatial memory impaired by irradiation, assessed in Morris water maze. A significant correlation between the number of proliferative cells and cognitive performances was found, too. Our study indicates that melatonin may decrease the loss of hippocampal neurons in the CA1 region and improve cognitive abilities after irradiation.
Collapse
Affiliation(s)
- Natália Pipová Kokošová
- Department of Animal Physiology, Faculty of Science, Institute of Biology and Ecology, P. J. Šafárik University in Košice, Košice, Slovak Republic
| | - Terézia Kisková
- Department of Animal Physiology, Faculty of Science, Institute of Biology and Ecology, P. J. Šafárik University in Košice, Košice, Slovak Republic
| | - Katarína Vilhanová
- Department of Animal Physiology, Faculty of Science, Institute of Biology and Ecology, P. J. Šafárik University in Košice, Košice, Slovak Republic
| | - Andrea Štafuriková
- Department of Animal Physiology, Faculty of Science, Institute of Biology and Ecology, P. J. Šafárik University in Košice, Košice, Slovak Republic
| | - Rastislav Jendželovský
- Department of Cell Biology, Faculty of Science, Institute of Biology and Ecology, P. J. Šafárik University in Košice, Košice, Slovak Republic
| | - Enikő Račeková
- Institute of Neurobiology, Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic
| | - Beňadik Šmajda
- Department of Animal Physiology, Faculty of Science, Institute of Biology and Ecology, P. J. Šafárik University in Košice, Košice, Slovak Republic
| |
Collapse
|
16
|
Liu B, Hinshaw RG, Le KX, Park MA, Wang S, Belanger AP, Dubey S, Frost JL, Shi Q, Holton P, Trojanczyk L, Reiser V, Jones PA, Trigg W, Di Carli MF, Lorello P, Caldarone BJ, Williams JP, O'Banion MK, Lemere CA. Space-like 56Fe irradiation manifests mild, early sex-specific behavioral and neuropathological changes in wildtype and Alzheimer's-like transgenic mice. Sci Rep 2019; 9:12118. [PMID: 31431669 PMCID: PMC6702228 DOI: 10.1038/s41598-019-48615-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
Space travel will expose people to high-energy, heavy particle radiation, and the cognitive deficits induced by this exposure are not well understood. To investigate the short-term effects of space radiation, we irradiated 4-month-old Alzheimer’s disease (AD)-like transgenic (Tg) mice and wildtype (WT) littermates with a single, whole-body dose of 10 or 50 cGy 56Fe ions (1 GeV/u) at Brookhaven National Laboratory. At ~1.5 months post irradiation, behavioural testing showed sex-, genotype-, and dose-dependent changes in locomotor activity, contextual fear conditioning, grip strength, and motor learning, mainly in Tg but not WT mice. There was little change in general health, depression, or anxiety. Two months post irradiation, microPET imaging of the stable binding of a translocator protein ligand suggested no radiation-specific change in neuroinflammation, although initial uptake was reduced in female mice independently of cerebral blood flow. Biochemical and immunohistochemical analyses revealed that radiation reduced cerebral amyloid-β levels and microglia activation in female Tg mice, modestly increased microhemorrhages in 50 cGy irradiated male WT mice, and did not affect synaptic marker levels compared to sham controls. Taken together, we show specific short-term changes in neuropathology and behaviour induced by 56Fe irradiation, possibly having implications for long-term space travel.
Collapse
Affiliation(s)
- Bin Liu
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA
| | - Robert G Hinshaw
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Kevin X Le
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mi-Ae Park
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Shuyan Wang
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Anthony P Belanger
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Shipra Dubey
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Jeffrey L Frost
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Qiaoqiao Shi
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA.,Harvard Medical School, Boston, MA, 02115, USA
| | - Peter Holton
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Lee Trojanczyk
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | | | - Paul A Jones
- GE Healthcare, Chalfont St Giles, HP8 4SP, United Kingdom
| | - William Trigg
- GE Healthcare, Chalfont St Giles, HP8 4SP, United Kingdom
| | - Marcelo F Di Carli
- Harvard Medical School, Boston, MA, 02115, USA.,Department of Radiology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Paul Lorello
- Harvard Medical School Mouse Behavior Core, Boston, MA, 02115, USA
| | | | - Jacqueline P Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Cynthia A Lemere
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, 02115, USA. .,Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
17
|
Jiang C, Stewart LT, Kuo HC, McGilberry W, Wall SB, Liang B, van Groen T, Bailey SM, Kim YI, Tipple TE, Jones DP, McMahon LL, Liu RM. Cyclic O 3 exposure synergizes with aging leading to memory impairment in male APOE ε3, but not APOE ε4, targeted replacement mice. Neurobiol Aging 2019; 81:9-21. [PMID: 31207469 DOI: 10.1016/j.neurobiolaging.2019.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/25/2019] [Accepted: 05/09/2019] [Indexed: 10/26/2022]
Abstract
The etiology of late-onset Alzheimer's disease is unknown. Recent epidemiological studies suggest that exposure to high levels of ozone (O3) may be a risk factor for late-onset Alzheimer's disease. Nonetheless, whether and how O3 exposure contributes to AD development remains to be determined. In this study, we tested the hypothesis that O3 exposure synergizes with the genetic risk factor APOE ε4 and aging leading to AD, using male apolipoprotein E (apoE)4 and apoE3 targeted replacement mice as men have increased risk exposure to high levels of O3 via working environments and few studies have addressed APOE ε4 effects on males. Surprisingly, our results show that O3 exposure impairs memory in old apoE3, but not old apoE4 or young apoE3 and apoE4, male mice. Further studies show that old apoE4 mice have increased hippocampal activities or expression of some enzymes involved in antioxidant defense, diminished protein oxidative modification, and neuroinflammation following O3 exposure compared with old apoE3 mice. These novel findings highlight the complexity of interactions between APOE genotype, age, and environmental exposure in AD development.
Collapse
Affiliation(s)
- Chunsun Jiang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Luke T Stewart
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hui-Chien Kuo
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William McGilberry
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie B Wall
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bill Liang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Thomas van Groen
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Young-Il Kim
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Trent E Tipple
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dean P Jones
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lori L McMahon
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rui-Ming Liu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
18
|
Female mice are protected from space radiation-induced maladaptive responses. Brain Behav Immun 2018; 74:106-120. [PMID: 30107198 PMCID: PMC8715721 DOI: 10.1016/j.bbi.2018.08.008] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/01/2018] [Accepted: 08/03/2018] [Indexed: 01/15/2023] Open
Abstract
Interplanetary exploration will be humankind's most ambitious expedition and the journey required to do so, is as intimidating as it is intrepid. One major obstacle for successful deep space travel is the possible negative effects of galactic cosmic radiation (GCR) exposure. Here, we investigate for the first time how combined GCR impacts long-term behavioral and cellular responses in male and female mice. We find that a single exposure to simulated GCR induces long-term cognitive and behavioral deficits only in the male cohorts. GCR exposed male animals have diminished social interaction, increased anxiety-like phenotype and impaired recognition memory. Remarkably, we find that the female cohorts did not display any cognitive or behavioral deficits after GCR exposure. Mechanistically, the maladaptive behavioral responses observed only in the male cohorts correspond with microglia activation and synaptic loss in the hippocampus, a brain region involved in the cognitive domains reported here. Furthermore, we measured reductions in AMPA expressing synaptic terminals in the hippocampus. No changes in any of the molecular markers measured here are observed in the females. Taken together these findings suggest that GCR exposure can regulate microglia activity and alter synaptic architecture, which in turn leads to a range of cognitive alterations in a sex dependent manner. These results identify sex-dependent differences in behavioral and cognitive domains revealing promising cellular and molecular intervention targets to reduce GCR-induced chronic cognitive deficits thereby boosting chances of success for humans in deep space missions such as the upcoming Mars voyage.
Collapse
|
19
|
Boehm-Cagan A, Bar R, Liraz O, Bielicki JK, Johansson JO, Michaelson DM. ABCA1 Agonist Reverses the ApoE4-Driven Cognitive and Brain Pathologies. J Alzheimers Dis 2018; 54:1219-1233. [PMID: 27567858 DOI: 10.3233/jad-160467] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The allele ɛ4 of apolipoprotein E (apoE4) is the most prevalent genetic risk factor for Alzheimer's disease (AD) and is therefore a promising therapeutic target. Human and animal model studies suggest that apoE4 is hypolipidated; accordingly, we have previously shown that the retinoid X receptor (RXR) agonist bexarotene upregulates ABCA1, the main apoE-lipidating protein, resulting in increased lipidation of apoE4, and the subsequent reversal of the pathological effects of apoE4, namely: accumulation of Aβ42 and hyperphosphorylated tau, as well as reduction in the levels of synaptic markers and cognitive deficits. Since the RXR system has numerous other targets, it is important to devise the means of activating ABCA1 selectively. We presently utilized CS-6253, a peptide shown to directly activate ABCA1 in vitro, and examined the extent to which it can affect the degree of lipidation of apoE4 in vivo and counteract the associated brain and behavioral pathologies. This revealed that treatment of young apoE4-targeted replacement mice with CS-6253 increases the lipidation of apoE4. This was associated with a reversal of the apoE4-driven Aβ42 accumulation and tau hyperphosphorylation in hippocampal neurons, as well as of the synaptic impairments and cognitive deficits. These findings suggest that the pathological effects of apoE4 in vivo are associated with decreased activation of ABCA1 and impaired lipidation of apoE4 and that the downstream brain-related pathology and cognitive deficits can be counteracted by treatment with the ABCA1 agonist CS-6253. These findings have important clinical ramifications and put forward ABCA1 as a promising target for apoE4-related treatment of AD.
Collapse
Affiliation(s)
- Anat Boehm-Cagan
- The Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Roni Bar
- The Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ori Liraz
- The Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - John K Bielicki
- Life Sciences Division, Lawrence Berkeley National Laboratory, University of California, Berkeley, CA, USA
| | | | - Daniel M Michaelson
- The Department of Neurobiology, The George S. Wise Faculty of Life Sciences, The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Fu Z, Zhao Y, Zhang K, Wang J, Zhang M, Zhao X. Age-Dependent Responses of Brain Myelin Integrity and Behavioral Performance to Radiation in Mice. Radiat Res 2017; 188:505-516. [PMID: 28937316 DOI: 10.1667/rr14732.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation therapy is widely used to treat primary and metastatic brain tumors, but it may also lead to delayed neurological complications. Oligodendrocytes in the central nervous system produce myelin, and myelin integrity becomes highly vulnerable after brain irradiation. In this study, mice at different developmental stages were used to test the age-dependent sensitivity of myelin formation and maintenance, as well as behavioral performance after whole-brain irradiation (WBI). Mice at postnatal days 21 and 28 and at 2 months received a single dose of 25 Gy WBI. Behavioral tests for general locomotor activity and motor coordination revealed an age-dependent response after WBI. Quantitative observation revealed a sharp decrease in the number of oligodendrocytes beginning at day 1 after WBI, which recovered during different observation intervals in white matter and gray matter in mice of different ages. Myelin basic protein (MBP) staining revealed disparate quantities in an age- and brain-region-dependent pattern between groups after WBI, which was confirmed using Black-Gold staining. In summary, the response to radiation in mice of different ages provided insight into the potential of oligogenesis in microenvironments at respective stages of myelin regeneration, which may reduce central nervous system impairment and optimize the prognosis after radiation treatment.
Collapse
Affiliation(s)
- Zhimeng Fu
- a Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032 China
| | - Yunfei Zhao
- a Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032 China.,b 93514 Hospital, Tangshan 064200 China
| | - Kaixiang Zhang
- a Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032 China
| | - Jian Wang
- a Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032 China
| | - Min Zhang
- a Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032 China
| | - Xianghui Zhao
- a Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic Medicine, Fourth Military Medical University, Xi'an 710032 China
| |
Collapse
|
21
|
Thomas R, Morris AW, Tai LM. Epidermal growth factor prevents APOE4-induced cognitive and cerebrovascular deficits in female mice. Heliyon 2017; 3:e00319. [PMID: 28626809 PMCID: PMC5463012 DOI: 10.1016/j.heliyon.2017.e00319] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/15/2017] [Accepted: 05/31/2017] [Indexed: 02/07/2023] Open
Abstract
Cerebrovascular dysfunction is re-emerging as a major component of aging, and may contribute to the risk of developing Alzheimer's disease (AD). Two important risk factors for cerebrovascular dysfunction are APOE and female sex, which are primarily researched in the context of high amyloid-β (Aβ) levels as found in AD. However, APOE4 and sex modulate Aβ-independent pathways that may induce cerebrovascular dysfunction as a downstream consequence. Therefore, testing the activity of factors that target cerebrovascular dysfunction in Aβ-independent models that incorporate APOE4 and female sex is crucial. We have previously demonstrated that peripheral administration of the epidermal growth factor (EGF) prevents cognitive dysfunction, cerebrovascular leakiness, and cerebrovascular coverage deficits in female mice that express APOE4 and overproduce Aβ, without affecting Aβ levels. These data raise the question of whether EGF protects the cerebrovasculature from general stress-induced damage. Therefore, the goal of this study was to determine whether EGF prevents Aβ-independent cerebrovascular dysfunction. In eight-month old mice that express human APOE, the interaction of APOE4 and female sex induced cognitive dysfunction, increased cerebrovascular leakiness and lowered vessel coverage. Importantly, in a prevention paradigm (from six to eight and a half months of age), EGF ameliorated cognitive decline and cerebrovascular deficits in female mice that express APOE4. Thus, developing treatment strategies based on EGF signaling could provide alternative therapeutic options for age-related cerebrovascular dysfunction and reduce AD risk.
Collapse
Affiliation(s)
| | | | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
22
|
Thomas R, Zuchowska P, Morris AWJ, Marottoli FM, Sunny S, Deaton R, Gann PH, Tai LM. Epidermal growth factor prevents APOE4 and amyloid-beta-induced cognitive and cerebrovascular deficits in female mice. Acta Neuropathol Commun 2016; 4:111. [PMID: 27788676 PMCID: PMC5084423 DOI: 10.1186/s40478-016-0387-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 10/22/2016] [Indexed: 11/10/2022] Open
Abstract
Cerebrovascular (CV) dysfunction is emerging as a critical component of Alzheimer's disease (AD), including altered CV coverage. Angiogenic growth factors (AGFs) are key for controlling CV coverage, especially during disease pathology. Therefore, evaluating the effects of AGFs in vivo can provide important information on the role of CV coverage in AD. We recently demonstrated that epidermal growth factor (EGF) prevents amyloid-beta (Aβ)-induced damage to brain endothelial cells in vitro. Here, our goal was to assess the protective effects of EGF on cognition, CV coverage and Aβ levels using an AD-Tg model that incorporates CV relevant AD risk factors. APOE4 is the greatest genetic risk factor for sporadic AD especially in women and is associated with CV dysfunction. EFAD mice express human APOE3 (E3FAD) or APOE4 (E4FAD), overproduce human Aβ42 and are a well characterized model of APOE pathology. Thus, initially the role of APOE and sex in cognitive and CV dysfunction was assessed in EFAD mice in order to identify a group for EGF treatment. At 8 months E4FAD female mice were cognitively impaired, had low CV coverage, high microbleeds and low plasma EGF levels. Therefore, E4FAD female mice were selected for an EGF prevention paradigm (300 μg/kg/wk, 6 to 8.5 months). EGF prevented cognitive decline and was associated with lower microbleeds and higher CV coverage, but not changes in Aβ levels. Collectively, these data suggest that EGF can prevent Aβ-induced damage to the CV. Developing therapeutic strategies based on AGFs may be particularly efficacious for APOE4-induced AD risk.
Collapse
Affiliation(s)
- Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Paulina Zuchowska
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Alan W. J. Morris
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Felecia M. Marottoli
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Sangeeta Sunny
- Research Histology and Tissue Imaging Core (RHTIC), University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Ryan Deaton
- Research Histology and Tissue Imaging Core (RHTIC), University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Peter H. Gann
- Research Histology and Tissue Imaging Core (RHTIC), University of Illinois at Chicago, Chicago, IL 60612 USA
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612 USA
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612 USA
| |
Collapse
|
23
|
Effects of Aging on Hippocampal Neurogenesis After Irradiation. Int J Radiat Oncol Biol Phys 2016; 94:1181-9. [DOI: 10.1016/j.ijrobp.2015.12.364] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 12/16/2015] [Accepted: 12/21/2015] [Indexed: 12/11/2022]
|
24
|
Kugelman T, Zuloaga DG, Weber S, Raber J. Post-training gamma irradiation-enhanced contextual fear memory associated with reduced neuronal activation of the infralimbic cortex. Behav Brain Res 2015; 298:1-11. [PMID: 26522840 DOI: 10.1016/j.bbr.2015.10.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/21/2015] [Accepted: 10/25/2015] [Indexed: 01/07/2023]
Abstract
The brain might be exposed to irradiation under a variety of situations, including clinical treatments, nuclear accidents, dirty bomb scenarios, and military and space missions. Correctly recalling tasks learned prior to irradiation is important but little is known about post-learning effects of irradiation. It is not clear whether exposure to X-ray irradiation during memory consolidation, a few hours following training, is associated with altered contextual fear conditioning 24h after irradiation and which brain region(s) might be involved in these effects. Brain immunoreactivity patterns of the immediately early gene c-Fos, a marker of cellular activity was used to determine which brain areas might be altered in post-training irradiation memory retention tasks. In this study, we show that post-training gamma irradiation exposure (1 Gy) enhanced contextual fear memory 24h later and is associated with reduced cellular activation in the infralimbic cortex. Reduced GABA-ergic neurotransmission in parvalbumin-positive cells in the infralimbic cortex might play a role in this post-training radiation-enhanced contextual fear memory.
Collapse
Affiliation(s)
- Tara Kugelman
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | - Damian G Zuloaga
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | - Sydney Weber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA; Oregon Health and Science University, Portland, OR 97239, USA; Division of Neuroscience, ONPRC, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
25
|
Kokošová N, Tomášová L, Kisková T, Šmajda B. Neuronal analysis and behaviour in prenatally gamma-irradiated rats. Cell Mol Neurobiol 2015; 35:45-55. [PMID: 25537960 PMCID: PMC11486342 DOI: 10.1007/s10571-014-0144-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 11/19/2014] [Indexed: 01/09/2023]
Abstract
The intrauterinal development in mammals represents a very sensitive period of life in relation to many environmental factors, including ionizing radiation (IR). The developing nervous system is particularly vulnerable to IR, and the consequences of exposure are of importance because of its potential health risks. The aim of our work was to assess whether prenatal irradiation of rats on the 17th day of embryonic development with a dose of 1 Gy would affect the formation of new cells and the number of mature neurons in the hippocampus and the selected forms of behaviour in the postnatal period. Male progeny of irradiated and control females was tested at ages of 3 weeks, 2 and 3 months. The number of mitotically active cells in the gyrus dentatus (GD) of the hippocampus was significantly reduced in irradiated rats aged 3 weeks. In irradiated rats aged 2 months, a significant reduction of mature neurons in CA1 area and in GD of the hippocampus was observed. The IR negatively influenced the spatial memory in Morris water maze, significantly decreased the exploratory behaviour and increased the anxiety-like behaviour in elevated plus-maze in rats aged 2 months. No significant differences were observed in animals aged 3 months compared with controls of the same age. A significant correlation between the number of mature neurons in the hilus and of the cognitive performances was found. Our results show that a low dose of radiation applied during the sensitive phase of brain development can influence the level of neurogenesis in the subgranular zone of GD and cause an impairment of the postnatal development of mental functions.
Collapse
Affiliation(s)
- Natália Kokošová
- Department of Animal Physiology, Faculty of Science, Institute of Biology and Ecology, P.J.Šafárik University, Šrobárova 2, 040 01, Košice, Slovak Republic,
| | | | | | | |
Collapse
|
26
|
Reverte I, Domingo JL, Colomina MT. Neurodevelopmental effects of decabromodiphenyl ether (BDE-209) in APOE transgenic mice. Neurotoxicol Teratol 2014; 46:10-7. [DOI: 10.1016/j.ntt.2014.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 11/26/2022]
|
27
|
Zou Y, Leu D, Chui J, Fike JR, Huang TT. Effects of altered levels of extracellular superoxide dismutase and irradiation on hippocampal neurogenesis in female mice. Int J Radiat Oncol Biol Phys 2013; 87:777-84. [PMID: 24064316 DOI: 10.1016/j.ijrobp.2013.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Indexed: 11/15/2022]
Abstract
PURPOSE Altered levels of extracellular superoxide dismutase (EC-SOD) and cranial irradiation have been shown to affect hippocampal neurogenesis. However, previous studies were only conducted in male mice, and it was not clear if there was a difference between males and females. Therefore, female mice were studied and the results compared with those generated in male mice from an earlier study. METHODS AND MATERIALS Female wild-type, EC-SOD-null (KO), and EC-SOD bigenic mice with neuronal-specific expression of EC-SOD (OE) were subjected to a single dose of 5-Gy gamma rays to the head at 8 weeks of age. Progenitor cell proliferation, differentiation, and long-term survival of newborn neurons were determined. RESULTS Similar to results from male mice, EC-SOD deficiency and irradiation both resulted in significant reductions in mature newborn neurons in female mice. EC-SOD deficiency reduced long-term survival of newborn neurons whereas irradiation reduced progenitor cell proliferation. Overexpression of EC-SOD corrected the negative impacts from EC-SOD deficiency and irradiation and normalized the production of newborn neurons in OE mice. Expression of neurotrophic factors brain-derived neurotrophic factor and neurotrophin-3 were significantly reduced by irradiation in wild-type mice, but the levels were not changed in KO and OE mice even though both cohorts started out with a lower baseline level. CONCLUSION In terms of hippocampal neurogenesis, EC-SOD deficiency and irradiation have the same overall effects in males and females at the age the studies were conducted.
Collapse
Affiliation(s)
- Yani Zou
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, California
| | | | | | | | | |
Collapse
|
28
|
Allen AR, Eilertson K, Sharma S, Schneider D, Baure J, Allen B, Rosi S, Raber J, Fike JR. Effects of radiation combined injury on hippocampal function are modulated in mice deficient in chemokine receptor 2 (CCR2). Radiat Res 2013; 180:78-88. [PMID: 23772926 DOI: 10.1667/rr3344.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Chemokines and their receptors play a crucial role in normal brain function as well as in pathological conditions such as injury and disease-associated neuroinflammation. Chemokine receptor-2 (CCR2), which mediates the recruitment of infiltrating and resident microglia to sites of central nervous system (CNS) inflammation, is upregulated by ionizing irradiation and traumatic brain injury. Our objective was to determine if a deficiency in CCR2 and subsequent effects on brain microglia affect neurogenesis and cognitive function after radiation combined injury (RCI). CCR2 knock-out ⁻/⁻ and wild-type (WT) mice received 4 Gy of whole body ¹³⁷Cs irradiation. Immediately after irradiation, unilateral traumatic brain injury was induced using a controlled cortical impact system. Forty-four days postirradiation, animals were tested for hippocampus-dependent cognitive performance in the Morris water-maze. After cognitive testing, animals were euthanized and their brains snap frozen for immunohistochemical assessment of neuroinflammation (activated microglia) and neurogenesis in the hippocampal dentate gyrus. All animals were able to locate the visible and hidden platform locations in the water maze; however, treatment effects were seen when spatial memory retention was assessed in the probe trials (no platform). In WT animals that received combined injury, a significant impairment in spatial memory retention was observed in the probe trial after the first day of hidden platform training (first probe trial). This impairment was associated with increased neurogenesis in the ipsilateral hemisphere of the dentate gyrus. In contrast, CCR2⁻/⁻ mice, independent of insult showed significant memory retention in the first probe trial and there were no differences in the numbers of newly born neurons in the animals receiving irradiation, trauma or combined injury. Although the mechanisms involved are not clear, our data suggests that CCR2 deficiency can exert a protective effect preventing the impairment of cognitive function after combined injury.
Collapse
Affiliation(s)
- Antiño R Allen
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California, San Francisco, California 94110, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Olsen RHJ, Agam M, Davis MJ, Raber J. ApoE isoform-dependent deficits in extinction of contextual fear conditioning. GENES, BRAIN, AND BEHAVIOR 2012; 11:806-12. [PMID: 22883220 PMCID: PMC3642038 DOI: 10.1111/j.1601-183x.2012.00833.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/03/2012] [Accepted: 08/02/2012] [Indexed: 11/26/2022]
Abstract
The three major human apoE isoforms (apoE2, apoE3 and apoE4) are encoded by distinct alleles (ϵ2, ϵ3 and ϵ4). Compared with ϵ3, ϵ4 is associated with increased risk to develop Alzheimer's disease (AD), cognitive impairments in Parkinson's disease (PD), and other conditions. In contrast, a recent study indicated an increased susceptibility to the recurring and re-experiencing symptom cluster of Post-Traumatic Stress Disorder (PTSD), as well as related memory impairments, in patients carrying at least one ϵ2 allele. Contextual fear conditioning and extinction are used in human and animal models to study this symptom cluster. In this study, acquisition (day 1, training), consolidation (day 2, first day of re-exposure) and extinction (days 2-5) of conditioned contextual fear in human apoE2, apoE3 and apoE4 targeted replacement and C57BL/6J wild-type (WT) mice was investigated. Male and female apoE2 showed acquisition and retrieval of conditioned fear, but failed to exhibit extinction. In contrast, WT, apoE3 and apoE4 mice showed extinction. While apoE2 mice exhibited lower freezing in response to the context on day 2 than apoE3 and apoE4 mice, this cannot explain their extinction deficit as WT mice exhibited similar freezing levels as apoE2 mice on day 2 but still exhibited extinction. Elevating freezing through extended training preserved extinction in controls, but failed to ameliorate extinction deficits in apoE2 animals. These data along with clinical data showing an association of apoE2 with susceptibility to specific symptom clusters in PTSD supports an important role for apoE isoform in the extinction of conditioned fear.
Collapse
Affiliation(s)
- Reid H J Olsen
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239
| | - Mati Agam
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239
| | - Matthew James Davis
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239
- Division of Neuroscience, Oregon National Primate Research
| |
Collapse
|
30
|
Roughton K, Kalm M, Blomgren K. Sex-dependent differences in behavior and hippocampal neurogenesis after irradiation to the young mouse brain. Eur J Neurosci 2012; 36:2763-72. [DOI: 10.1111/j.1460-9568.2012.08197.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
31
|
Beydoun MA, Boueiz A, Abougergi MS, Kitner-Triolo MH, Beydoun HA, Resnick SM, O'Brien R, Zonderman AB. Sex differences in the association of the apolipoprotein E epsilon 4 allele with incidence of dementia, cognitive impairment, and decline. Neurobiol Aging 2012; 33:720-731.e4. [PMID: 20619505 PMCID: PMC2974952 DOI: 10.1016/j.neurobiolaging.2010.05.017] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Revised: 05/12/2010] [Accepted: 05/16/2010] [Indexed: 11/20/2022]
Abstract
We examined longitudinal associations between the apolipoprotein E ε4 allele (ApoE4(+) status) and several cognitive outcomes and tested effect modification by sex. Data on 644 non-Hispanic Caucasian adults, from the Baltimore Longitudinal Study of Aging (BLSA) were used. Dementia onset, cognitive impairment and decline were assessed longitudinally. After 27.5 years median follow-up, 113 participants developed dementia. ApoE4(+) predicted dementia significantly (hazard ratio [HR] = 2.89; 95% confidence interval [CI], 1.93-4.33), with nonsignificant sex differences. Taking all time points for predicting cognition, women had significantly stronger positive associations than men between ApoE4(+) status and impairment or decline on the California Verbal Learning Test (CVLT; delayed recall and List A total recall) and on Verbal Fluency Test-Categories. This ApoE4 × sex interaction remained significant with Bonferroni correction only for CVLT-delayed recall. Taking time points prior to dementia for cognitive predictions, the positive association between impairment in CVLT-delayed recall and ApoE4(+) status remained stronger among women, though only before Bonferroni correction. While ApoE4(+) status appears to be a sex neutral risk factor for dementia, its association with verbal memory and learning decline and impairment was stronger among women.
Collapse
Affiliation(s)
- May A Beydoun
- National Institute on Aging, NIA/NIH/IRP, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Haley GE, Villasana L, Dayger C, Davis MJ, Raber J. Apolipoprotein e genotype-dependent paradoxical short-term effects of (56)fe irradiation on the brain. Int J Radiat Oncol Biol Phys 2012; 84:793-9. [PMID: 22401921 DOI: 10.1016/j.ijrobp.2011.12.049] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 12/07/2011] [Accepted: 12/12/2011] [Indexed: 10/28/2022]
Abstract
PURPOSE In humans, apolipoprotein E (apoE) is encoded by three major alleles (ε2, ε3, and ε4) and, compared to apoE3, apoE4 increases the risk of developing Alzheimer disease and cognitive impairments following various environmental challenges. Exposure to irradiation, including that of (56)Fe, during space missions poses a significant risk to the central nervous system, and apoE isoform might modulate this risk. METHODS AND MATERIALS We investigated whether apoE isoform modulates hippocampus-dependent cognitive performance starting 2 weeks after (56)Fe irradiation. Changes in reactive oxygen species (ROS) can affect cognition and are induced by irradiation. Therefore, after cognitive testing, we assessed hippocampal ROS levels in ex vivo brain slices, using the ROS-sensitive fluorescent probe, dihydroethidium (DHE). Brain levels of 3-nitrotyrosine (3-NT), CuZn superoxide dismutase (CuZnSOD), extracellular SOD, and apoE were assessed using Western blotting analysis. RESULTS In the water maze, spatial memory retention was impaired by irradiation in apoE2 and apoE4 mice but enhanced by irradiation in apoE3 mice. Irradiation reduced DHE-oxidation levels in the enclosed blade of the dentate gyrus and levels of 3-NT and CuZnSOD in apoE2 but not apoE3 or apoE4 mice. Finally, irradiation increased apoE levels in apoE3 but not apoE2 or apoE4 mice. CONCLUSIONS The short-term effects of (56)Fe irradiation on hippocampal ROS levels and hippocampus-dependent spatial memory retention are apoE isoform-dependent.
Collapse
Affiliation(s)
- Gwendolen E Haley
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | | | | | |
Collapse
|
33
|
Siegel JA, Haley GE, Raber J. Apolipoprotein E isoform-dependent effects on anxiety and cognition in female TR mice. Neurobiol Aging 2012; 33:345-58. [PMID: 20400205 PMCID: PMC2935518 DOI: 10.1016/j.neurobiolaging.2010.03.002] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 10/30/2009] [Accepted: 03/02/2010] [Indexed: 10/19/2022]
Abstract
Compared with apoE3, apoE4 is associated with increased risk to develop age-related cognitive decline, particularly in women. In this study, young, middle-aged, and old female mice expressing human apoE under control of the mouse apoE promoter were behaviorally analyzed. Cognitive performance in the water maze decreased with age in all mice. Compared with apoE2 and apoE3 mice, apoE4 mice showed better cognitive performance and higher measures of anxiety than apoE2 and apoE3 mice. Measures of anxiety correlated with cognitive performance in the water maze and passive avoidance tests and might have contributed to the enhanced cognitive performance of the apoE4 mice. ApoE4 mice showed better water maze learning and higher cortical apoE levels than mice expressing apoE4 in astrocytes under control of the GFAP promoter. This was not seen in apoE3 mice. There were no line differences in either genotype in spatial memory retention in the probe trial following the last day of hidden platform training. Thus, the promoter used to express apoE4 critically modulates its effects on brain function.
Collapse
Affiliation(s)
- Jessica A. Siegel
- Department of Behavioral Neuroscience, Oregon Health and Science University, 8131 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Gwendolen E. Haley
- Department of Behavioral Neuroscience, Oregon Health and Science University, 8131 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, 8131 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Department of Neurology, Oregon Health and Science University, 8131 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Division of Neuroscience ONPRC, Oregon Health and Science University, 8131 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
34
|
Rosi S, Ferguson R, Fishman K, Allen A, Raber J, Fike JR. The polyamine inhibitor alpha-difluoromethylornithine modulates hippocampus-dependent function after single and combined injuries. PLoS One 2012; 7:e31094. [PMID: 22299052 PMCID: PMC3267765 DOI: 10.1371/journal.pone.0031094] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 01/02/2012] [Indexed: 12/31/2022] Open
Abstract
Exposure to uncontrolled irradiation in a radiologic terrorism scenario, a natural disaster or a nuclear battlefield, will likely be concomitantly superimposed on other types of injury, such as trauma. In the central nervous system, radiation combined injury (RCI) involving irradiation and traumatic brain injury may have a multifaceted character. This may entail cellular and molecular changes that are associated with cognitive performance, including changes in neurogenesis and the expression of the plasticity-related immediate early gene Arc. Because traumatic stimuli initiate a characteristic early increase in polyamine metabolism, we hypothesized that treatment with the polyamine inhibitor alpha-difluoromethylornithine (DFMO) would reduce the adverse effects of single or combined injury on hippocampus structure and function. Hippocampal dependent cognitive impairments were quantified with the Morris water maze and showed that DFMO effectively reversed cognitive impairments after all injuries, particularly traumatic brain injury. Similar results were seen with respect to the expression of Arc protein, but not neurogenesis. Given that polyamines have been found to modulate inflammatory responses in the brain we also assessed the numbers of total and newly born activated microglia, and found reduced numbers of newly born cells. While the mechanisms responsible for the improvement in cognition after DFMO treatment are not yet clear, the present study provides new and compelling data regarding the potential use of DFMO as a potential countermeasure against the adverse effects of single or combined injury.
Collapse
Affiliation(s)
- Susanna Rosi
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Ryan Ferguson
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Physical Therapy and Rehabilitation Science, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Kelly Fishman
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Antino Allen
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health and Science University, Portland, Oregon, United States of America
- Division of Neuroscience, Oregon National Primate Research Center (ONPRC), Oregon Health and Science University, Portland, Oregon, United States of America
| | - John R. Fike
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Radiation Oncology, University of California San Francisco, San Francisco, California, United States of America
- Brain and Spinal Injury Center, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
35
|
Conner KR, Forbes ME, Lee WH, Lee YW, Riddle DR. AT1 receptor antagonism does not influence early radiation-induced changes in microglial activation or neurogenesis in the normal rat brain. Radiat Res 2011; 176:71-83. [PMID: 21545290 PMCID: PMC3159150 DOI: 10.1667/rr2560.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Blockers of the renin-angiotensin-aldosterone system (RAAS) ameliorate cognitive deficits and some aspects of brain injury after whole-brain irradiation. We investigated whether treatment with the angiotensin II type 1 receptor antagonist L-158,809 at a dose that protects cognitive function after fractionated whole-brain irradiation reduced radiation-induced neuroinflammation and changes in hippocampal neurogenesis, well-characterized effects that are associated with radiation-induced brain injury. Male F344 rats received L-158,809 before, during and after a single 10-Gy dose of radiation. Expression of cytokines, angiotensin II receptors and angiotensin-converting enzyme 2 was evaluated by real-time PCR 24 h, 1 week and 12 weeks after irradiation. At the latter times, microglial density and proliferating and activated microglia were analyzed in the dentate gyrus of the hippocampus. Cell proliferation and neurogenesis were also quantified in the dentate subgranular zone. L-158,809 treatment modestly increased mRNA expression for Ang II receptors and TNF-α but had no effect on radiation-induced effects on hippocampal microglia or neurogenesis. Thus, although L-158,809 ameliorates cognitive deficits after whole-brain irradiation, the drug did not mitigate the neuroinflammatory microglial response or rescue neurogenesis. Additional studies are required to elucidate other mechanisms of normal tissue injury that may be modulated by RAAS blockers.
Collapse
Affiliation(s)
- Kelly R. Conner
- Program in Neuroscience, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - M. Elizabeth Forbes
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Won Hee Lee
- School of Biomedical Engineering and Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - Yong Woo Lee
- School of Biomedical Engineering and Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia
| | - David R. Riddle
- Program in Neuroscience, Wake Forest School of Medicine, Winston-Salem, North Carolina
- Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
36
|
Villasana LE, Benice TS, Raber J. Long-Term Effects of 56Fe Irradiation on Spatial Memory of Mice: Role of Sex and Apolipoprotein E Isoform. Int J Radiat Oncol Biol Phys 2011; 80:567-73. [DOI: 10.1016/j.ijrobp.2010.12.034] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2010] [Revised: 12/09/2010] [Accepted: 12/15/2010] [Indexed: 12/15/2022]
|
37
|
Dayger C, Villasana L, Pfankuch T, Davis M, Raber J. Effects of the SARM ACP-105 on rotorod performance and cued fear conditioning in sham-irradiated and irradiated female mice. Brain Res 2011; 1381:134-40. [PMID: 21219889 PMCID: PMC3048897 DOI: 10.1016/j.brainres.2010.12.088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/29/2010] [Accepted: 12/30/2010] [Indexed: 12/11/2022]
Abstract
Female mice are more susceptible to radiation-induced cognitive changes than male mice. Previously, we showed that, in female mice, androgens antagonize age-related cognitive decline in aged wild-type mice and androgens and selective androgen receptor modulators (SARMs) antagonize cognitive changes induced by human apolipoprotein E4, a risk factor for developing age-related cognitive decline. In this study, the potential effects of the SARM ACP-105 were assessed in female mice that were either sham-irradiated or irradiated with ¹³⁷Cesium at a dose of 10Gy. Behavioral testing started 2 weeks following irradiation. Irradiation impaired sensorimotor function in vehicle-treated mice but not in ACP-105-treated mice. Irradiation impaired cued fear conditioning and ACP-105 enhanced fear conditioning in sham-irradiated and irradiated mice. When immunoreactivity for microtubule-associated protein 2 was assessed in the cortex of sham-irradiated mice, there was a brain area × ACP-105 interaction. While ACP-105 reduced MAP-2 immunoreactivity in the sensorimotor cortex, there was a trend towards increased MAP-2 immunoreactivity in the enthorhinal cortex. No effect on MAP-2 immunoreactivity was seen in the irradiated cortex or sham-irradiated or irradiated hippocampus. Thus, there are relatively early radiation-induced behavioral changes in female mice and reduced MAP-2 levels in the sensorimotor cortex following ACP-105 treatment might contribute to enhanced rotorod performance.
Collapse
Affiliation(s)
- Catherine Dayger
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Laura Villasana
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Timothy Pfankuch
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Matthew Davis
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon 97239
- Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239
- Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
38
|
Raber J, Villasana L, Rosenberg J, Zou Y, Huang TT, Fike JR. Irradiation enhances hippocampus-dependent cognition in mice deficient in extracellular superoxide dismutase. Hippocampus 2011; 21:72-80. [PMID: 20020436 PMCID: PMC2891276 DOI: 10.1002/hipo.20724] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The effects of ionizing irradiation on the brain are associated with oxidative stress. While oxidative stress following irradiation is generally viewed as detrimental for hippocampal function, it might have beneficial effects as part of an adaptive or preconditioning response to a subsequent challenge. Here we show that in contrast to what is seen in wild-type mice, irradiation enhances hippocampus- dependent cognitive measures in mice lacking extracellular superoxide dismutase. These outcomes were associated with genotype-dependent effects on measures of oxidative stress. When cortices and hippocampi were analyzed for nitrotyrosine formation as an index of oxidative stress, the levels were chronically elevated in mice lacking extracellular superoxide dismutase. However, irradiation caused a greater increase in nitrotyrosine levels in wild-type mice than mice lacking extracellular superoxide dismutase. These paradoxical genotype-dependent effects of irradiation on measures of oxidative stress and cognitive function underscore potential beneficial effects associated with chronic oxidative stress if it exists prior to a secondary insult such as irradiation.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | | | | | | | | | | |
Collapse
|
39
|
Rosi S, Belarbi K, Ferguson RA, Fishman K, Obenaus A, Raber J, Fike JR. Trauma-induced alterations in cognition and Arc expression are reduced by previous exposure to 56Fe irradiation. Hippocampus 2010; 22:544-54. [PMID: 21192069 DOI: 10.1002/hipo.20920] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/23/2010] [Indexed: 12/31/2022]
Abstract
Exposure to ionizing irradiation may affect brain functions directly, but may also change tissue sensitivity to a secondary insult such as trauma, stroke, or degenerative disease. To determine if a low dose of particulate irradiation sensitizes the brain to a subsequent injury, C56BL6 mice were exposed to brain only irradiation with 0.5 Gy of (56) Fe ions. Two months later, unilateral traumatic brain injury was induced using a controlled cortical impact system. Three weeks after trauma, animals received multiple BrdU injections and 30 days later were tested for cognitive performance in the Morris water maze. All animals were able to locate the visible and hidden platform during training; however, treatment effects were seen when spatial memory retention was assessed in the probe trial (no platform). Although sham and irradiated animals showed spatial memory retention, mice that received trauma alone did not. When trauma was preceded by irradiation, performance in the water maze was not different from sham-treated animals, suggesting that low-dose irradiation had a protective effect in the context of a subsequent traumatic injury. Measures of hippocampal neurogenesis showed that combined injury did not induce any changes greater that those seen after trauma or radiation alone. After trauma, there was a significant decrease in the percentage of neurons expressing the behaviorally induced immediate early gene Arc in both hemispheres, without associated neuronal loss. After combined injury there were no differences relative to sham-treated mice. Our results suggest that combined injury resulted in decreased alterations of our endpoints compared to trauma alone. Although the underlying mechanisms are not yet known, these results resemble a preconditioning, adaptive, or inducible-like protective response, where a sublethal or potentially injurious stimulus (i.e., irradiation) induces tolerance to a subsequent and potentially more damaging insult (trauma).
Collapse
Affiliation(s)
- Susanna Rosi
- Brain and Spinal Injury Center, University of California, San Francisco, California 94110, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Robbins ME, Zhao W, Garcia-Espinosa MA, Diz DI. Renin-angiotensin system blockers and modulation of radiation-induced brain injury. Curr Drug Targets 2010; 11:1413-22. [PMID: 20583976 PMCID: PMC3068470 DOI: 10.2174/1389450111009011413] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2009] [Accepted: 04/05/2010] [Indexed: 01/05/2023]
Abstract
Radiation-induced brain injury remains a major cause of morbidity in cancer patients with primary or metastatic brain tumors. Approximately 200,000 individuals/year are treated with fractionated partial or whole-brain irradiation, and > half will survive long enough (≤6 months) to develop radiation-induced brain injury, including cognitive impairment. Although short-term treatments have shown efficacy, no long-term treatments or preventive approaches are presently available for modulating radiation-induced brain injury. Based on previous preclinical studies clearly demonstrating that renin-angiotensin system (RAS) blockers can modulate radiation-induced late effects in the kidney and lung, we and others hypothesized that RAS blockade would similarly modulate radiation-induced brain injury. Indeed, studies in the last 5 years have shown that both angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin II type 1 receptor antagonists (AT(1)RAs) can prevent/ameliorate radiation-induced brain injury, including cognitive impairment, in the rat. The mechanistic basis for this RAS blocker-mediated effect remains the subject of ongoing investigations. Putative mechanisms include, i] blockade of Ang II/NADPH oxidase-mediated oxidative stress and neuroinflammation, and ii] a change in the balance of angiotensin (Ang) peptides from the pro-inflammatory and pro-oxidative Ang II to the anti-inflammatory and anti-oxidative Ang-1-7). However, given that both ACEIs and AT(1)RAs are 1] well-tolerated drugs routinely prescribed for hypertension, 2] exhibit some antitumor properties, and 3] can prevent/ameliorate radiation-induced brain injury, they appear to be ideal drugs for future clinical trials, offering the promise of improving the quality of life of brain tumor patients receiving brain irradiation.
Collapse
Affiliation(s)
- M E Robbins
- Department of Radiation Oncology, Brain Tumor Center of Excellence, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | | | | | |
Collapse
|
41
|
Villasana L, Pfankuch T, Raber J. Isoform-dependent effects of apoE on doublecortin-positive cells and microtubule-associated protein 2 immunoreactivity following (137)Cs irradiation. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2010; 49:421-426. [PMID: 20458592 DOI: 10.1007/s00411-010-0290-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2009] [Accepted: 04/19/2010] [Indexed: 05/29/2023]
Abstract
Previously we found apoE isoform-dependent effects of (137)Cs irradiation on cognitive function of female mice 3 months following irradiation. Alterations in the number of immature neurons and in the levels of the dendritic marker microtubule-associated protein 2 (MAP-2) might contribute to the cognitive changes following irradiation. Therefore, in the present study we determined if, following (137)Cs irradiation, there are apoE isoform-dependent effects on loss of doublecortin-positive neuroprogenitor cells or MAP-2 immumonoreactivity. In the dentate gyrus, CA1 and CA3 regions of the hippocampus, enthorhinal and sensorimotor cortex, and central and basolateral nuclei of the amygdala of apoE3 female mice, MAP-2 immunoreactivity increased 3 months following (137)Cs irradiation. In addition, at 8 h following irradiation, the number of doublecortin-positive cells was higher in apoE3 than apoE2 or apoE4 mice. Together, these data indicate that brains of apoE3 mice respond differently to (137)Cs irradiation than those of apoE2 or apoE4 mice.
Collapse
Affiliation(s)
- Laura Villasana
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR 97239, USA
| | | | | |
Collapse
|
42
|
Knapman A, Heinzmann JM, Hellweg R, Holsboer F, Landgraf R, Touma C. Increased stress reactivity is associated with cognitive deficits and decreased hippocampal brain-derived neurotrophic factor in a mouse model of affective disorders. J Psychiatr Res 2010; 44:566-75. [PMID: 20035953 DOI: 10.1016/j.jpsychires.2009.11.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2009] [Revised: 11/10/2009] [Accepted: 11/20/2009] [Indexed: 01/08/2023]
Abstract
Cognitive deficits are a common feature of major depression (MD), with largely unknown biological underpinnings. In addition to the affective and cognitive symptoms of MD, a dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis is commonly observed in these patients. Increased plasma glucocorticoid levels are known to render the hippocampus susceptible to neuronal damage. This structure is important for learning and memory, creating a potential link between HPA axis dysregulation and cognitive deficits in depression. In order to further elucidate how altered stress responsiveness may contribute to the etiology of MD, three mouse lines with high (HR), intermediate (IR), or low (LR) stress reactivity were generated by selective breeding. The aim of the present study was to investigate whether increased stress reactivity is associated with deficits in hippocampus-dependent memory tests. To this end, we subjected mice from the HR, IR, and LR breeding lines to tests of recognition memory, spatial memory, and depression-like behavior. In addition, measurements of brain-derived neurotrophic factor (BDNF) in the hippocampus and plasma of these animals were conducted. Our results demonstrate that HR mice exhibit hippocampus-dependent memory deficits along with decreased hippocampal, but not plasma, BDNF levels. Thus, the stress reactivity mouse lines are a promising animal model of the cognitive deficits in MD with the unique feature of a genetic predisposition for an altered HPA axis reactivity, which provides the opportunity to explore the progression of the symptoms of MD, predisposing genetic factors as well as new treatment strategies.
Collapse
Affiliation(s)
- A Knapman
- Department of Behavioral Neuroendocrinology, Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Benice TS, Raber J. Castration and training in a spatial task alter the number of immature neurons in the hippocampus of male mice. Brain Res 2010; 1329:21-9. [PMID: 20233585 PMCID: PMC2857657 DOI: 10.1016/j.brainres.2010.03.028] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2009] [Revised: 02/26/2010] [Accepted: 03/06/2010] [Indexed: 11/18/2022]
Abstract
New neurons are generated in the granule cell layer of the dentate gyrus (GCL) throughout adulthood. This process is modulated by many environmental and neurochemical factors. We previously observed that castrated mice, compared to sham-operated mice, perform poorly in the delayed matching to place water-maze task (DMTP). In this study, we quantified the number of doublecortin expressing (DCX+) immature neurons and Ki-67 expressing (Ki-67+) proliferating progenitors in mice previously tested in a spatial DMTP task, a nonspatial DMTP, or that received equivalent amounts of handling only. Regardless of DMTP training experience, castration reduced immature neuron number in the GCL but had no effect on proliferating progenitors. Compared to handling only, visible DMTP training reduced the immature neuron number, but hidden DMTP training had no effect. Castration did not alter these environmental effects. Finally, performance on the spatial DMTP task did not correlate with immature neuron number. In addition, while the number of immature neurons was strongly reduced following cranial irradiation with (137)Cs, this treatment did not affect spatial DMTP performance. Thus, in mice, castration disrupts spatial memory and reduces immature neuron number, but there is no strong link between these effects.
Collapse
Affiliation(s)
- Ted S. Benice
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR 97239
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239
- Division of Neuroscience ONPRC, Oregon Health & Science University, Portland, OR 97239
| |
Collapse
|
44
|
Duvoisin RM, Villasana L, Pfankuch T, Raber J. Sex-dependent cognitive phenotype of mice lacking mGluR8. Behav Brain Res 2010; 209:21-6. [PMID: 20080129 DOI: 10.1016/j.bbr.2010.01.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 01/06/2010] [Accepted: 01/07/2010] [Indexed: 11/27/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) modulate glutamatergic and GABAergic neurotransmission. mGluR8 is generally located presynaptically where it regulates neurotransmitter release. Previously we reported that 6-month-old mGluR8(-/-) male mice show higher measures of anxiety in anxiety tests involving avoidable anxiety-provoking stimuli than age-matched wild-type male mice. In wild-type mice, middle-aged females and males show higher measures of anxiety in such tests and reduced spatial learning than young adults. In this study we evaluated in middle-aged mice the effects of mGluR8 deficiency on measures of anxiety involving avoidable and unavoidable anxiety-provoking stimuli and on cognitive performance and whether these effects are sex-dependent. Female and male mGluR8(-/-) mice showed increased measures of anxiety in the open field. In contrast, male mGluR8(-/-) mice showed increased but female mGluR8(-/-) mice decreased measures of anxiety in the elevated plus maze and the acoustic startle response. mGluR8 deficiency impaired novel location recognition and spatial memory retention in the water maze. The impairment in spatial memory retention in the water maze, but not in novel location recognition, was more pronounced in female than male mice. Thus, potential sex differences in the therapeutic effects of mGluR8 modulation to reduce measures of anxiety and improve cognitive performance should be carefully considered.
Collapse
Affiliation(s)
- Robert M Duvoisin
- Department of Physiology and Pharmacology, Oregon Health & Science University, USA
| | | | | | | |
Collapse
|
45
|
Villasana L, Rosenberg J, Raber J. Sex-dependent effects of 56Fe irradiation on contextual fear conditioning in C57BL/6J mice. Hippocampus 2010; 20:19-23. [PMID: 19489001 PMCID: PMC2850563 DOI: 10.1002/hipo.20659] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Effects of irradiation on hippocampal function have been mostly studied in male rodents and relatively little is known about potential effects of irradiation on hippocampal function in female rodents. Moreover, although the long-term effects of clinical radiation on cognitive function have been well established, the effects of other forms of irradiation, such as high charged, high energy radiation (HZE particles) that astronauts encounter during space missions have not been well characterized. In this study we compared the effects of (56)Fe irradiation on fear conditioning in C57BL/6J female and male mice. Hippocampus-dependent contextual fear conditioning was impaired in female mice but improved in male mice following (56)Fe irradiation. Such impairment was not seen for hippocampus-independent cued fear conditioning. Thus, the effects of (56)Fe irradiation on hippocampus-dependent contextual fear conditioning are critically modulated by sex.
Collapse
Affiliation(s)
- Laura Villasana
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon, USA
| | | | | |
Collapse
|
46
|
Jazin E, Cahill L. Sex differences in molecular neuroscience: from fruit flies to humans. Nat Rev Neurosci 2010; 11:9-17. [DOI: 10.1038/nrn2754] [Citation(s) in RCA: 177] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
47
|
Li G, Bien-Ly N, Andrews-Zwilling Y, Xu Q, Bernardo A, Ring K, Halabisky B, Deng C, Mahley RW, Huang Y. GABAergic interneuron dysfunction impairs hippocampal neurogenesis in adult apolipoprotein E4 knockin mice. Cell Stem Cell 2009; 5:634-45. [PMID: 19951691 PMCID: PMC2992822 DOI: 10.1016/j.stem.2009.10.015] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2009] [Revised: 10/09/2009] [Accepted: 10/14/2009] [Indexed: 02/02/2023]
Abstract
Apolipoprotein (apo) E, a polymorphic protein with three isoforms (apoE2, apoE3, and apoE4), is essential for lipid homeostasis. Carriers of apoE4 are at higher risk for developing Alzheimer's disease. We have investigated adult neurogenesis in mice with knockout (KO) for apoE or with knockin (KI) alleles for human apoE3 or apoE4, and we report that neurogenesis is reduced in both apoE-KO and apoE4-KI mice. In apoE-KO mice, increased BMP signaling promoted glial differentiation at the expense of neurogenesis. In contrast, in apoE4-KI mice, presynaptic GABAergic input-mediated maturation of newborn neurons was diminished. Tau phosphorylation, an Alzheimer's disease characteristic, and levels of neurotoxic apoE fragments were both elevated in apoE4-KI hippocampal neurons concomitant with decreased GABAergic interneuron survival. Potentiating GABAergic signaling restored neuronal maturation and neurogenesis in apoE4-KI mice to normal levels. These findings suggest that GABAergic signaling can be targeted to mitigate the deleterious effects of apoE4 on neurogenesis.
Collapse
Affiliation(s)
- Gang Li
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
- Department of Neurology, University of California, San Francisco, California 94143, USA
| | - Nga Bien-Ly
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, California 94143, USA
| | - Yaisa Andrews-Zwilling
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
- Department of Neurology, University of California, San Francisco, California 94143, USA
| | - Qin Xu
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
- Gladstone Institute of Cardiovascular Disease, San Francisco, California 94158, USA
| | - Aubrey Bernardo
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
| | - Karen Ring
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, California 94143, USA
| | - Brian Halabisky
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
| | - Changhui Deng
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California 94143, USA
| | - Robert W. Mahley
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
- Gladstone Institute of Cardiovascular Disease, San Francisco, California 94158, USA
- Department of Medicine, University of California, San Francisco, California 94143, USA
- Department of Pathology, University of California, San Francisco, California 94143, USA
| | - Yadong Huang
- Gladstone Institute of Neurological Disease, San Francisco, California 94158, USA
- Gladstone Institute of Cardiovascular Disease, San Francisco, California 94158, USA
- Biomedical Sciences Graduate Program, University of California, San Francisco, California 94143, USA
- Department of Pathology, University of California, San Francisco, California 94143, USA
- Department of Neurology, University of California, San Francisco, California 94143, USA
| |
Collapse
|
48
|
Fishman K, Baure J, Zou Y, Huang TT, Andres-Mach M, Rola R, Suarez T, Acharya M, Limoli CL, Lamborn KR, Fike JR. Radiation-induced reductions in neurogenesis are ameliorated in mice deficient in CuZnSOD or MnSOD. Free Radic Biol Med 2009; 47:1459-67. [PMID: 19703553 PMCID: PMC2767469 DOI: 10.1016/j.freeradbiomed.2009.08.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 07/30/2009] [Accepted: 08/19/2009] [Indexed: 12/13/2022]
Abstract
Ionizing irradiation significantly affects hippocampal neurogenesis and is associated with cognitive impairments; these effects may be influenced by an altered microenvironment. Oxidative stress is a factor that has been shown to affect neurogenesis, and one of the protective pathways that deal with such stress involves the antioxidant enzyme superoxide dismutase (SOD). This study addressed what impact a deficiency in cytoplasmic (SOD1) or mitochondrial (SOD2) SOD has on radiation effects on hippocampal neurogenesis. Wild-type (WT) and SOD1 and SOD2 knockout (KO) mice received a single X-ray dose of 5 Gy, and quantification of the survival and phenotypic fate of newly generated cells in the dentate subgranular zone was performed 2 months later. Radiation exposure reduced neurogenesis in WT mice but had no apparent effect in KO mice, although baseline levels of neurogenesis were reduced in both SOD KO strains before irradiation. Additionally, there were marked and significant differences between WT and both KO strains in how irradiation affected newly generated astrocytes and activated microglia. The mechanism(s) responsible for these effects is not yet known, but a pilot in vitro study suggests a "protective" effect of elevated levels of superoxide. Overall, these data suggest that under conditions of SOD deficiency, there is a common pathway dictating how neurogenesis is affected by ionizing irradiation.
Collapse
Affiliation(s)
- Kelly Fishman
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Jennifer Baure
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Yani Zou
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Ting-Ting Huang
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
- GRECC, VA Palo Alto Health Care System, Palo Alto, CA, USA
| | - Marta Andres-Mach
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Radoslaw Rola
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, F. Skubiszewski Medical University, Lublin, Poland
| | - Tatiana Suarez
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Munjal Acharya
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Kathleen R. Lamborn
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - John R. Fike
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
49
|
Abstract
This review summarizes some of the topics discussed at the 28th Annual Symposium of the Society of Toxicologic Pathology. The symposium was held in Washington, DC, in 2009 and dealt with unintended adverse events associated with cranial irradiation as part of cancer therapy. We will discuss the importance of considering genetic susceptibility and sex differences in susceptibility to develop these adverse events. Further, we will discuss potential mechanisms contributing to these events, including alterations in neurogenesis and increased oxidative stress following irradiation and potential alterations in synaptic and dendritic markers.
Collapse
Affiliation(s)
- Jacob Raber
- Departments of Behavioral Neuroscience and of Neurology,
and Division of Neuroscience, ONPRC, Oregon Health and Science University,
Portland, Oregon, USA,
| |
Collapse
|
50
|
Reduced levels of human apoE4 protein in an animal model of cognitive impairment. Neurobiol Aging 2009; 32:791-801. [PMID: 19577821 DOI: 10.1016/j.neurobiolaging.2009.05.011] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2009] [Revised: 05/08/2009] [Accepted: 05/14/2009] [Indexed: 11/22/2022]
Abstract
The APOE4 allele is the most common genetic determinant for Alzheimer's disease (AD) in the developed world. APOE genotype specific differences in brain apolipoprotein E protein levels have been observed in numerous studies since the discovery of APOE4's link to AD. Since the human apoE4 targeted replacement mice display characteristics of cognitive impairment we sought to determine if reduced levels of apoE might provide one explanation for this impairment. We developed a novel mass spectrometry method to measure apoE protein levels in plasma. Additionally, we developed an ELISA that replicates the mass spectrometry data and enables the rapid quantitation of apoE in plasma, brain and cerebrospinal fluid. We detected a significant decrease in plasma, brain and cerebrospinal fluid apoE levels in the apoE4 mice compared to apoE2 and E3 mice. We also measured a small (∼19%) decrease in brain apoE levels from aged, non-demented APOE4 carriers. Our findings suggest that a fraction of APOE4-linked AD may be due to insufficient levels of functional apoE required to maintain neuronal health.
Collapse
|